Human Perivascular Stem Cell-Derived Extracellular Vesicles Mediate Bone

Total Page:16

File Type:pdf, Size:1020Kb

Human Perivascular Stem Cell-Derived Extracellular Vesicles Mediate Bone RESEARCH ARTICLE Human perivascular stem cell-derived extracellular vesicles mediate bone repair Jiajia Xu1, Yiyun Wang1, Ching-Yun Hsu1, Yongxing Gao1, Carolyn Ann Meyers1, Leslie Chang1, Leititia Zhang1,2, Kristen Broderick3, Catherine Ding4, Bruno Peault4,5,6, Kenneth Witwer7,8, Aaron Watkins James1* 1Department of Pathology, Johns Hopkins University, Baltimore, United States; 2Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, China; 3Department of Surgery, Johns Hopkins University, Baltimore, United States; 4Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, UCLA, Orthopaedic Hospital, Los Angeles, United States; 5Centre For Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; 6MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom; 7Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, United States; 8Department of Neurology, Johns Hopkins University, Baltimore, United States Abstract The vascular wall is a source of progenitor cells that are able to induce skeletal repair, primarily by paracrine mechanisms. Here, the paracrine role of extracellular vesicles (EVs) in bone healing was investigated. First, purified human perivascular stem cells (PSCs) were observed to induce mitogenic, pro-migratory, and pro-osteogenic effects on osteoprogenitor cells while in non- contact co-culture via elaboration of EVs. PSC-derived EVs shared mitogenic, pro-migratory, and pro-osteogenic properties of their parent cell. PSC-EV effects were dependent on surface- associated tetraspanins, as demonstrated by EV trypsinization, or neutralizing antibodies for CD9 or CD81. Moreover, shRNA knockdown in recipient cells demonstrated requirement for the CD9/ CD81 binding partners IGSF8 and PTGFRN for EV bioactivity. Finally, PSC-EVs stimulated bone repair, and did so via stimulation of skeletal cell proliferation, migration, and osteodifferentiation. *For correspondence: In sum, PSC-EVs mediate the same tissue repair effects of perivascular stem cells, and represent an [email protected] ‘off-the-shelf’ alternative for bone tissue regeneration. Competing interest: See DOI: https://doi.org/10.7554/eLife.48191.001 page 17 Funding: See page 17 Received: 03 May 2019 Accepted: 03 September 2019 Introduction Published: 04 September 2019 Stromal progenitor cells within vessel walls have multipotent properties (Cathery et al., 2018; Corselli et al., 2012; Covas et al., 2008; Crisan et al., 2008; Dellavalle et al., 2007; Farrington- Reviewing editor: Clifford J Rock et al., 2004), are native forerunners of mesenchymal stem cells (MSCs), and participate in Rosen, Maine Medical Center endogenous bone repair (Diaz-Flores et al., 1991; Diaz-Flores et al., 1992; Grcevic et al., 2012). Research Institute, United States When purified based on expression of CD146 (Mel-CAM) and CD34, human perivascular stem cells Copyright Xu et al. This article (PSCs) from adipose tissue or other tissue compartments speed bone repair (Askarinam et al., is distributed under the terms of 2013; Chung et al., 2014; James et al., 2017a; James et al., 2012a; James et al., 2012b; the Creative Commons Tawonsawatruk et al., 2016). Although direct incorporation of human PSCs into chondroblasts, Attribution License, which permits unrestricted use and osteoblasts and osteocytes occurs (James et al., 2012b), PSCs induce bone healing either primarily redistribution provided that the or exclusively via paracrine stimulation of the resident host cells within the defect niche original author and source are (Chung et al., 2014; Tawonsawatruk et al., 2016). High expression and secretion of osteoinductive credited. proteins has been observed among freshly sorted or culture-expanded PSCs, including bone Xu et al. eLife 2019;8:e48191. DOI: https://doi.org/10.7554/eLife.48191 1 of 23 Research article Cell Biology Stem Cells and Regenerative Medicine eLife digest Throughout our lives, our bodies need to heal after injury. Blood vessels are found throughout the body’s tissues and are a source of cells that guide the process of repair. These cells, called perivascular stem cells (PSCs), are a type of stem cell found in the lining of blood vessels. Stem cells are cells that can become one of several different types of mature cells, depending on what the body needs. Extracellular vesicles are bundles of chemical signals that cells send into their external environment. Just like an address or a tag on a parcel, specific molecules mark the exterior surface of these bundles to deliver the message to the right recipient. Stem cells often use extracellular vesicles to communicate with surrounding cells. One role of PSCs is repairing damage to bones. Unusually, they do not turn into new bone cells and so do not directly contribute to the re-growing tissue. Instead, PSCs act indirectly, by stimulating the cells around them. How PSCs send these ‘repair instructions’ has, however, remained unclear. Xu et al. wanted to determine if PSCs used extracellular vesicles to direct bone repair, and if so, what ‘tags’ needed to be on the vesicles and on the receiving cells for this to happen. Experiments using PSCs and immature bone cells grown in the laboratory allowed the PSCs’ effect on bone cells to be simulated in a Petri dish. The two types of cells were grown on either side of a barrier, which separated them physically but allowed chemical signals through. In response to the PSCs, the immature bone cells multiplied, started to move (which is something they need to do to heal damaged tissue), and began to resemble mature bone cells. Analysis of the signals released by the PSCs revealed that these were indeed extracellular vesicles, and that they were tagged by specific proteins called tetraspanins. Genetic manipulation of the immature bone cells later showed that these cells needed specific ‘receiver’ molecules to respond to the PSCs. Adding only extracellular vesicles to the bone cells, without any PSCs, confirmed that it was indeed the vesicles that triggered the healing response. Finally, giving the vesicles to mice with bone damage helped them to heal faster than untreated animals. These results have uncovered a key mechanism by which stem cells control the repair of bone tissue. This could one day lead to better treatments for patients recovering from fractures or needing bone surgery. DOI: https://doi.org/10.7554/eLife.48191.002 morphogenetic proteins (BMPs) and vascular endothelial growth factor (VEGF) among others (Chen et al., 2009; Hardy et al., 2017; James et al., 2017b). However, the exact paracrine intermediaries of PSC-induced bone defect healing are not known. Extracellular vesicles (EVs), including exosomes and microvesicles, carry a repertoire of bioactive molecules: proteins, nucleic acids, lipids and carbohydrates (Colombo et al., 2014; Cvjetkovic et al., 2016; Mateescu et al., 2017). The heterogenous nature of EVs is well described, with EVs of a diameter <100–150 nm and multivesicular body (MVB) origin termed exosomes (Gould and Raposo, 2013; Mateescu et al., 2017). Stromal progenitor cells elaborate EVs, and EVs have been observed to harbor many of the regenerative properties of their parent cell (Lakhter and Sims, 2015). In bone repair, CD9 knockout mice, which have impaired EV elaboration, have delayed appendicular bone healing, that may be rescued by mesenchymal progenitor cell-derived EVs (Furuta et al., 2016). As well, induced pluripotent stem cell-derived EVs have been shown to incite osteogenesis and vasculogenesis in vivo (Qin et al., 2016; Todorova et al., 2017). The potential role of miRNA cargo in EVs has been implicated in these effects, however the mechanisms that underlie EV-mediated reparative effects in bone are essentially unknown. Here, we observe that in similarity to their parent cell, PSC-derived EVs incite pleiotropic effects on skeletal progenitor cells, including mitogenic, pro-migratory, pro-osteogenic effects, and broad ranging changes in the cellular transcriptome of the recipient cell. These bioactive effects require EV tetraspanin activity, as well as expression of their binding partners IGSF8 and PTGFRN by the recipi- ent progenitor cell. These cellular effects of PSC-EVs on skeletal progenitor cells converge to incite intramembranous bone repair in a mouse model. Xu et al. eLife 2019;8:e48191. DOI: https://doi.org/10.7554/eLife.48191 2 of 23 Research article Cell Biology Stem Cells and Regenerative Medicine Results Perivascular stem cells induce paracrine effects on BMSCs via EV release Perivascular stem/stromal cells (PSCs) were derived from human white adipose tissue using previ- ously reported methods, by FACS selection of the related populations of CD34+CD146-CD45-CD31- adventitial progenitor cells and CD146+CD34-CD45-CD31- pericytes (Corselli et al., 2012; Crisan et al., 2008; James et al., 2017a; Meyers et al., 2018aMeyers et al., 2018b; Meyers et al., 2018b)(Figure 1—figure supplement 1). Frequencies of human PSCs within adipose tissue are summarized in Supplementary file 2, and were within our previously reported ranges (James et al., 2012a; West et al., 2016). In order to better understand the paracrine effects of PSCs in bone repair, we first defined a set of replicable in vitro paracrine effects of human PSCs on recipient human BMSCs (Figure 1). Prior to all experiments, cell surface antigens of human PSCs and BMSCs were typified by flow cytometry, and the multilineage differentiation
Recommended publications
  • Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse
    Welcome to More Choice CD Marker Handbook For more information, please visit: Human bdbiosciences.com/eu/go/humancdmarkers Mouse bdbiosciences.com/eu/go/mousecdmarkers Human and Mouse CD Marker Handbook Human and Mouse CD Marker Key Markers - Human Key Markers - Mouse CD3 CD3 CD (cluster of differentiation) molecules are cell surface markers T Cell CD4 CD4 useful for the identification and characterization of leukocytes. The CD CD8 CD8 nomenclature was developed and is maintained through the HLDA (Human Leukocyte Differentiation Antigens) workshop started in 1982. CD45R/B220 CD19 CD19 The goal is to provide standardization of monoclonal antibodies to B Cell CD20 CD22 (B cell activation marker) human antigens across laboratories. To characterize or “workshop” the antibodies, multiple laboratories carry out blind analyses of antibodies. These results independently validate antibody specificity. CD11c CD11c Dendritic Cell CD123 CD123 While the CD nomenclature has been developed for use with human antigens, it is applied to corresponding mouse antigens as well as antigens from other species. However, the mouse and other species NK Cell CD56 CD335 (NKp46) antibodies are not tested by HLDA. Human CD markers were reviewed by the HLDA. New CD markers Stem Cell/ CD34 CD34 were established at the HLDA9 meeting held in Barcelona in 2010. For Precursor hematopoetic stem cell only hematopoetic stem cell only additional information and CD markers please visit www.hcdm.org. Macrophage/ CD14 CD11b/ Mac-1 Monocyte CD33 Ly-71 (F4/80) CD66b Granulocyte CD66b Gr-1/Ly6G Ly6C CD41 CD41 CD61 (Integrin b3) CD61 Platelet CD9 CD62 CD62P (activated platelets) CD235a CD235a Erythrocyte Ter-119 CD146 MECA-32 CD106 CD146 Endothelial Cell CD31 CD62E (activated endothelial cells) Epithelial Cell CD236 CD326 (EPCAM1) For Research Use Only.
    [Show full text]
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Flow Reagents Single Color Antibodies CD Chart
    CD CHART CD N° Alternative Name CD N° Alternative Name CD N° Alternative Name Beckman Coulter Clone Beckman Coulter Clone Beckman Coulter Clone T Cells B Cells Granulocytes NK Cells Macrophages/Monocytes Platelets Erythrocytes Stem Cells Dendritic Cells Endothelial Cells Epithelial Cells T Cells B Cells Granulocytes NK Cells Macrophages/Monocytes Platelets Erythrocytes Stem Cells Dendritic Cells Endothelial Cells Epithelial Cells T Cells B Cells Granulocytes NK Cells Macrophages/Monocytes Platelets Erythrocytes Stem Cells Dendritic Cells Endothelial Cells Epithelial Cells CD1a T6, R4, HTA1 Act p n n p n n S l CD99 MIC2 gene product, E2 p p p CD223 LAG-3 (Lymphocyte activation gene 3) Act n Act p n CD1b R1 Act p n n p n n S CD99R restricted CD99 p p CD224 GGT (γ-glutamyl transferase) p p p p p p CD1c R7, M241 Act S n n p n n S l CD100 SEMA4D (semaphorin 4D) p Low p p p n n CD225 Leu13, interferon induced transmembrane protein 1 (IFITM1). p p p p p CD1d R3 Act S n n Low n n S Intest CD101 V7, P126 Act n p n p n n p CD226 DNAM-1, PTA-1 Act n Act Act Act n p n CD1e R2 n n n n S CD102 ICAM-2 (intercellular adhesion molecule-2) p p n p Folli p CD227 MUC1, mucin 1, episialin, PUM, PEM, EMA, DF3, H23 Act p CD2 T11; Tp50; sheep red blood cell (SRBC) receptor; LFA-2 p S n p n n l CD103 HML-1 (human mucosal lymphocytes antigen 1), integrin aE chain S n n n n n n n l CD228 Melanotransferrin (MT), p97 p p CD3 T3, CD3 complex p n n n n n n n n n l CD104 integrin b4 chain; TSP-1180 n n n n n n n p p CD229 Ly9, T-lymphocyte surface antigen p p n p n
    [Show full text]
  • Supp Material.Pdf
    Simon et al. Supplementary information: Table of contents p.1 Supplementary material and methods p.2-4 • PoIy(I)-poly(C) Treatment • Flow Cytometry and Immunohistochemistry • Western Blotting • Quantitative RT-PCR • Fluorescence In Situ Hybridization • RNA-Seq • Exome capture • Sequencing Supplementary Figures and Tables Suppl. items Description pages Figure 1 Inactivation of Ezh2 affects normal thymocyte development 5 Figure 2 Ezh2 mouse leukemias express cell surface T cell receptor 6 Figure 3 Expression of EZH2 and Hox genes in T-ALL 7 Figure 4 Additional mutation et deletion of chromatin modifiers in T-ALL 8 Figure 5 PRC2 expression and activity in human lymphoproliferative disease 9 Figure 6 PRC2 regulatory network (String analysis) 10 Table 1 Primers and probes for detection of PRC2 genes 11 Table 2 Patient and T-ALL characteristics 12 Table 3 Statistics of RNA and DNA sequencing 13 Table 4 Mutations found in human T-ALLs (see Fig. 3D and Suppl. Fig. 4) 14 Table 5 SNP populations in analyzed human T-ALL samples 15 Table 6 List of altered genes in T-ALL for DAVID analysis 20 Table 7 List of David functional clusters 31 Table 8 List of acquired SNP tested in normal non leukemic DNA 32 1 Simon et al. Supplementary Material and Methods PoIy(I)-poly(C) Treatment. pIpC (GE Healthcare Lifesciences) was dissolved in endotoxin-free D-PBS (Gibco) at a concentration of 2 mg/ml. Mice received four consecutive injections of 150 μg pIpC every other day. The day of the last pIpC injection was designated as day 0 of experiment.
    [Show full text]
  • CD Markers Are Routinely Used for the Immunophenotyping of Cells
    ptglab.com 1 CD MARKER ANTIBODIES www.ptglab.com Introduction The cluster of differentiation (abbreviated as CD) is a protocol used for the identification and investigation of cell surface molecules. So-called CD markers are routinely used for the immunophenotyping of cells. Despite this use, they are not limited to roles in the immune system and perform a variety of roles in cell differentiation, adhesion, migration, blood clotting, gamete fertilization, amino acid transport and apoptosis, among many others. As such, Proteintech’s mini catalog featuring its antibodies targeting CD markers is applicable to a wide range of research disciplines. PRODUCT FOCUS PECAM1 Platelet endothelial cell adhesion of blood vessels – making up a large portion molecule-1 (PECAM1), also known as cluster of its intracellular junctions. PECAM-1 is also CD Number of differentiation 31 (CD31), is a member of present on the surface of hematopoietic the immunoglobulin gene superfamily of cell cells and immune cells including platelets, CD31 adhesion molecules. It is highly expressed monocytes, neutrophils, natural killer cells, on the surface of the endothelium – the thin megakaryocytes and some types of T-cell. Catalog Number layer of endothelial cells lining the interior 11256-1-AP Type Rabbit Polyclonal Applications ELISA, FC, IF, IHC, IP, WB 16 Publications Immunohistochemical of paraffin-embedded Figure 1: Immunofluorescence staining human hepatocirrhosis using PECAM1, CD31 of PECAM1 (11256-1-AP), Alexa 488 goat antibody (11265-1-AP) at a dilution of 1:50 anti-rabbit (green), and smooth muscle KD/KO Validated (40x objective). alpha-actin (red), courtesy of Nicola Smart. PECAM1: Customer Testimonial Nicola Smart, a cardiovascular researcher “As you can see [the immunostaining] is and a group leader at the University of extremely clean and specific [and] displays Oxford, has said of the PECAM1 antibody strong intercellular junction expression, (11265-1-AP) that it “worked beautifully as expected for a cell adhesion molecule.” on every occasion I’ve tried it.” Proteintech thanks Dr.
    [Show full text]
  • Single-Cell Analysis Uncovers Fibroblast Heterogeneity
    ARTICLE https://doi.org/10.1038/s41467-020-17740-1 OPEN Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination ✉ Lars Muhl 1,2 , Guillem Genové 1,2, Stefanos Leptidis 1,2, Jianping Liu 1,2, Liqun He3,4, Giuseppe Mocci1,2, Ying Sun4, Sonja Gustafsson1,2, Byambajav Buyandelger1,2, Indira V. Chivukula1,2, Åsa Segerstolpe1,2,5, Elisabeth Raschperger1,2, Emil M. Hansson1,2, Johan L. M. Björkegren 1,2,6, Xiao-Rong Peng7, ✉ Michael Vanlandewijck1,2,4, Urban Lendahl1,8 & Christer Betsholtz 1,2,4 1234567890():,; Many important cell types in adult vertebrates have a mesenchymal origin, including fibro- blasts and vascular mural cells. Although their biological importance is undisputed, the level of mesenchymal cell heterogeneity within and between organs, while appreciated, has not been analyzed in detail. Here, we compare single-cell transcriptional profiles of fibroblasts and vascular mural cells across four murine muscular organs: heart, skeletal muscle, intestine and bladder. We reveal gene expression signatures that demarcate fibroblasts from mural cells and provide molecular signatures for cell subtype identification. We observe striking inter- and intra-organ heterogeneity amongst the fibroblasts, primarily reflecting differences in the expression of extracellular matrix components. Fibroblast subtypes localize to discrete anatomical positions offering novel predictions about physiological function(s) and regulatory signaling circuits. Our data shed new light on the diversity of poorly defined classes of cells and provide a foundation for improved understanding of their roles in physiological and pathological processes. 1 Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre, Blickagången 6, SE-14157 Huddinge, Sweden.
    [Show full text]
  • Attenuation of Pathogenic Immune Responses During Infection with Human and Simian Immunodeficiency Virus (HIV/SIV) by the Tetracycline Derivative Minocycline
    Attenuation of Pathogenic Immune Responses during Infection with Human and Simian Immunodeficiency Virus (HIV/SIV) by the Tetracycline Derivative Minocycline Julia L. Drewes1, Gregory L. Szeto1¤, Elizabeth L. Engle1, Zhaohao Liao1, Gene M. Shearer2,M. Christine Zink1*, David R. Graham1,3* 1 Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, 2 Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America, 3 Johns Hopkins Bayview Proteomics Center, Department of Medicine, Division of Cardiology, Johns Hopkins School of Medicine Bayview Campus, Baltimore, Maryland, United States of America Abstract HIV immune pathogenesis is postulated to involve two major mechanisms: 1) chronic innate immune responses that drive T cell activation and apoptosis and 2) induction of immune regulators that suppress T cell function and proliferation. Both arms are elevated chronically in lymphoid tissues of non-natural hosts, which ultimately develop AIDS. However, these mechanisms are not elevated chronically in natural hosts of SIV infection that avert immune pathogenesis despite similarly high viral loads. In this study we investigated whether minocycline could modulate these pathogenic antiviral responses in non-natural hosts of HIV and SIV. We found that minocycline attenuated in vitro induction of type I interferon (IFN) and the IFN-stimulated genes indoleamine 2,3-dioxygenase (IDO1) and TNF-related apoptosis inducing ligand (TRAIL) in human plasmacytoid dendritic cells and PBMCs exposed to aldrithiol-2 inactivated HIV or infectious influenza virus. Activation- induced TRAIL and expression of cytotoxic T-lymphocyte antigen 4 (CTLA-4) in isolated CD4+ T cells were also reduced by minocycline.
    [Show full text]
  • Genetic Defects in B-Cell Development and Their Clinical Consequences H Abolhassani,1,2 N Parvaneh,1 N Rezaei,1 L Hammarström,2 a Aghamohammadi1
    REVIEWS Genetic Defects in B-Cell Development and Their Clinical Consequences H Abolhassani,1,2 N Parvaneh,1 N Rezaei,1 L Hammarström,2 A Aghamohammadi1 1Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran 2Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden n Abstract Expression of selected genes in hematopoietic stem cells has been identified as a regulator of differentiation of B cells in the liver and bone marrow. Moreover, naïve B cells expressing surface immunoglobulin need other types of genes for antigen-dependent development in secondary lymphoid organs. Many advanced molecular mechanisms underlying primary antibody deficiencies in humans have been described. We provide an overview of the mutations in genes known to be involved in B-cell development and their clinical consequences. Key words: Genetic disorder. B-cell development. Primary antibody deficiencies. Clinical phenotypes. n Resumen Se ha identificado la expresión de genes seleccionados en las células pluripotenciales de médula ósea como reguladores de la diferenciación de las células B en el hígado y en médula ósea. Sin embargo, las células B naïve que expresan inmunoglubulinas de superficie, necesitan otros tipos de genes para su desarrollo en los órganos linfoides secundarios dependienteS de antígeno. Se han descrito muchos mecanismos moleculares avanzados que subrayan las inmunodeficiencias en humanos y esta revisión constituye una visión general de la mutación en todos los genes conocidos involucrados en el desarrollo de las células B y sus consecuencias clínicas. Palabras clave: Alteraciones genéticas. Desarrollo de las células B.
    [Show full text]
  • University of Groningen Mechanics of Extracellular Vesicles
    University of Groningen Mechanics of extracellular vesicles derived from malaria parasiteinfected Red Blood Cells Sorkin, Raya; Vorselen, Daan; Ofir-Birin, Yifat; Roos, Wouter H.; MacKintosh, Fred C.; Regev-Rudzki, Neta; Wuite, Gijs J. L. Published in: Journal of Extracellular Vesicles DOI: 10.3402/jev.v5.31552 IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below. Publication date: 2016 Link to publication in University of Groningen/UMCG research database Citation for published version (APA): Sorkin, R., Vorselen, D., Ofir-Birin, Y., Roos, W. H., MacKintosh, F. C., Regev-Rudzki, N., & Wuite, G. J. L. (2016). Mechanics of extracellular vesicles derived from malaria parasiteinfected Red Blood Cells. Journal of Extracellular Vesicles, 5, 86. https://doi.org/10.3402/jev.v5.31552 Copyright Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons). Take-down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum. Download date: 12-11-2019
    [Show full text]
  • Transcriptome Analysis Reveals Transmembrane Targets on Transplantable Midbrain Dopamine Progenitors
    Transcriptome analysis reveals transmembrane targets on transplantable midbrain dopamine progenitors Chris R. Byea, Marie E. Jönssonb, Anders Björklundb,1, Clare L. Parisha, and Lachlan H. Thompsona,1 aFlorey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia 3010; and bWallenberg Neuroscience Centre, Department of Experimental Medical Science, Lund University, S-22184 Lund, Sweden Contributed by Anders Björklund, February 5, 2015 (sent for review December 8, 2014; reviewed by Eva Hedlund and Tilo Kunath) An important challenge for the continued development of cell targeting of cell-specific surface antigens as a means to separate therapy for Parkinson’s disease (PD) is the establishment of proce- defined cell fractions from mixed populations. Central to the dures that better standardize cell preparations for use in transplan- development of the technique was the identification of trans- tation. Although cell sorting has been an anticipated strategy, its membrane proteins that selectively define a target cell population. + + application has been limited by lack of knowledge regarding trans- For example, isolation of CD34 and Thy1 cells facilitates puri- membrane proteins that can be used to target and isolate progen- fication of hematopoietic stem cells that can reconstitute the he- itors for midbrain dopamine (mDA) neurons. We used a “FACS-array” matopoietic system as part of treatment for cancer patients (7–10). approach to identify 18 genes for transmembrane proteins with A current obstacle for the development of similar techniques for high expression in mDA progenitors and describe the utility of four the purification of specific neural progenitor phenotypes, in- of these targets (Alcam, Chl1, Gfra1, and Igsf8) for isolating mDA cluding mDA progenitors, is a lack of knowledge regarding the progenitors from rat primary ventral mesencephalon through flow identity of suitable transmembrane targets.
    [Show full text]
  • Human CD Marker Chart Reviewed by HLDA1 Bdbiosciences.Com/Cdmarkers
    BD Biosciences Human CD Marker Chart Reviewed by HLDA1 bdbiosciences.com/cdmarkers 23-12399-01 CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD Alternative Name Ligands & Associated Molecules T Cell B Cell Dendritic Cell NK Cell Stem Cell/Precursor Macrophage/Monocyte Granulocyte Platelet Erythrocyte Endothelial Cell Epithelial Cell CD1a R4, T6, Leu6, HTA1 b-2-Microglobulin, CD74 + + + – + – – – CD93 C1QR1,C1qRP, MXRA4, C1qR(P), Dj737e23.1, GR11 – – – – – + + – – + – CD220 Insulin receptor (INSR), IR Insulin, IGF-2 + + + + + + + + + Insulin-like growth factor 1 receptor (IGF1R), IGF-1R, type I IGF receptor (IGF-IR), CD1b R1, T6m Leu6 b-2-Microglobulin + + + – + – – – CD94 KLRD1, Kp43 HLA class I, NKG2-A, p39 + – + – – – – – – CD221 Insulin-like growth factor 1 (IGF-I), IGF-II, Insulin JTK13 + + + + + + + + + CD1c M241, R7, T6, Leu6, BDCA1 b-2-Microglobulin + + + – + – – – CD178, FASLG, APO-1, FAS, TNFRSF6, CD95L, APT1LG1, APT1, FAS1, FASTM, CD95 CD178 (Fas ligand) + + + + + – – IGF-II, TGF-b latency-associated peptide (LAP), Proliferin, Prorenin, Plasminogen, ALPS1A, TNFSF6, FASL Cation-independent mannose-6-phosphate receptor (M6P-R, CIM6PR, CIMPR, CI- CD1d R3G1, R3 b-2-Microglobulin, MHC II CD222 Leukemia
    [Show full text]
  • Engineered Type 1 Regulatory T Cells Designed for Clinical Use Kill Primary
    ARTICLE Acute Myeloid Leukemia Engineered type 1 regulatory T cells designed Ferrata Storti Foundation for clinical use kill primary pediatric acute myeloid leukemia cells Brandon Cieniewicz,1* Molly Javier Uyeda,1,2* Ping (Pauline) Chen,1 Ece Canan Sayitoglu,1 Jeffrey Mao-Hwa Liu,1 Grazia Andolfi,3 Katharine Greenthal,1 Alice Bertaina,1,4 Silvia Gregori,3 Rosa Bacchetta,1,4 Norman James Lacayo,1 Alma-Martina Cepika1,4# and Maria Grazia Roncarolo1,2,4# Haematologica 2021 Volume 106(10):2588-2597 1Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 2Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 3San Raffaele Telethon Institute for Gene Therapy, Milan, Italy and 4Center for Definitive and Curative Medicine, Stanford School of Medicine, Stanford, CA, USA *BC and MJU contributed equally as co-first authors #AMC and MGR contributed equally as co-senior authors ABSTRACT ype 1 regulatory (Tr1) T cells induced by enforced expression of interleukin-10 (LV-10) are being developed as a novel treatment for Tchemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft-versus-host disease while mediating graft-versus-leukemia effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature.
    [Show full text]